14
Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis Brice Marcet a, , Jean-Marie Boeynaems a,b a Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Campus Erasme (Bât C5-110), route de Lennik 808, 1070 Brussels, Belgium b Department of Medical Chemistry, Erasme Hospital, Université Libre de Bruxelles, route de Lennik 808, 1070 Brussels, Belgium Abstract Cystic fibrosis (CF) is one of the most common lethal autosomal recessive genetic diseases in the Caucasian population, with a frequency of about 1 in 3000 livebirths. CF is due to a mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene encoding the CFTR protein, a cyclic adenosine 5-monophosphate (cAMP)-regulated chloride channel localized in the apical membrane of epithelial cells. CFTR is a multifunctional protein which, in addition to be a Cl-channel, is also a regulator of multiple ion channels and other proteins. In particular CFTR has been reported to play a role in the outflow of adenosine 5-triphosphate (ATP) from cells, but this remains controversial. Extracellular nucleotides are signaling molecules that regulate ion transport and mucociliary clearance by acting on P2 nucleotide receptors, in particular the P2Y 2 receptor. Nucleotides activating the P2Y 2 receptor represent thus one pharmacotherapeutic strategy to treat CF disease, via improvement of mucus hydration and mucociliary clearance in airways. Phase II clinical trials have recently shown that aerosolized denufosol (INS37217, Inspire®) improves pulmonary function in CF patients: denufosol was granted orphan drug status and phase III trials are planned. Here, we review what is known about the relationship between extracellular nucleotides and CFTR, the role of extracellular nucleotides in epithelial pathophysiology and their putative role as therapeutic agents. © 2006 Elsevier Inc. All rights reserved. Keywords: Cystic fibrosis; CFTR; Chloride channels; Extracellular nucleotides; P2Y receptors; Airway epithelium Abbreviations: ABC, adenosine 5-triphosphate-binding cassette; ADO, adenosine; ADP, adenosine 5-diphosphate; AEC, airway epithelial cell; AMP, adenosine 5- monophosphate; ASL, airway surface liquid; ATP, adenosine 5-triphosphate; ATPase, adenosine triphosphatase; CaCC, Ca 2+ -activated chloride channel; cAMP, cyclic adenosine 5-monophosphate; CF, cystic fibrosis; CFTR, cystic fibrosis transmembrane conductance regulator; CHO, Chinese hamster ovary; ClC, chloride channels; DC, dendritic cell; ENaC, epithelial sodium channel; IDO, indoleamine 2,3-dioxygenase; mRNA, messenger ribonucleic acid; NBD, nucleotide-binding domain; NSAP, nonspecific alkaline phosphatase; ORCC, outwardly rectifying chloride channel; PCL, periciliary liquid; PKA, protein kinase A; PKC, protein kinase C; RNA, ribonucleic acid; SLPI, secretory leukocyte protease inhibitor; UDP, uridine 5-diphosphate; UTP, uridine 5-triphosphate. Contents 1. Introduction................................................ 720 2. Does cystic fibrosis transmembrane conductance regulator control adenosine 5-triphosphate release? A controversial issue ...................................... 720 3. Role of P2 receptors in the regulation of cystic fibrosis transmembrane conductance regulator and other chloride channels .................................. 722 3.1. Cystic fibrosis transmembrane conductance regulator and P2Y 1 receptor ............. 722 3.2. Cystic fibrosis transmembrane conductance regulator, chloride channels and P2Y 2 receptors .. 722 3.3. Cystic fibrosis transmembrane conductance regulator, P2Y 4 and P2Y 6 receptors ......... 723 3.4. Cystic fibrosis transmembrane conductance regulator and P2X receptors ............. 724 Pharmacology & Therapeutics 112 (2006) 719 732 www.elsevier.com/locate/pharmthera The authors attest that there is no conflicts of interest. Corresponding author. E-mail addresses: [email protected], [email protected] (B. Marcet). 0163-7258/$ - see front matter © 2006 Elsevier Inc. All rights reserved. doi:10.1016/j.pharmthera.2006.05.010

Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis

Embed Size (px)

Citation preview

Page 1: Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis

s 112 (2006) 719–732www.elsevier.com/locate/pharmthera

Pharmacology & Therapeutic

Relationships between cystic fibrosis transmembrane conductance regulator,extracellular nucleotides and cystic fibrosis☆

Brice Marcet a,⁎, Jean-Marie Boeynaems a,b

a Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Campus Erasme (Bât C5-110), route de Lennik 808, 1070 Brussels, Belgiumb Department of Medical Chemistry, Erasme Hospital, Université Libre de Bruxelles, route de Lennik 808, 1070 Brussels, Belgium

Abstract

Cystic fibrosis (CF) is one of the most common lethal autosomal recessive genetic diseases in the Caucasian population, with a frequency ofabout 1 in 3000 livebirths. CF is due to a mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene encoding the CFTRprotein, a cyclic adenosine 5′-monophosphate (cAMP)-regulated chloride channel localized in the apical membrane of epithelial cells. CFTR is amultifunctional protein which, in addition to be a Cl-channel, is also a regulator of multiple ion channels and other proteins. In particular CFTRhas been reported to play a role in the outflow of adenosine 5′-triphosphate (ATP) from cells, but this remains controversial. Extracellularnucleotides are signaling molecules that regulate ion transport and mucociliary clearance by acting on P2 nucleotide receptors, in particular theP2Y2 receptor. Nucleotides activating the P2Y2 receptor represent thus one pharmacotherapeutic strategy to treat CF disease, via improvement ofmucus hydration and mucociliary clearance in airways. Phase II clinical trials have recently shown that aerosolized denufosol (INS37217,Inspire®) improves pulmonary function in CF patients: denufosol was granted orphan drug status and phase III trials are planned. Here, we reviewwhat is known about the relationship between extracellular nucleotides and CFTR, the role of extracellular nucleotides in epithelialpathophysiology and their putative role as therapeutic agents.© 2006 Elsevier Inc. All rights reserved.

Keywords: Cystic fibrosis; CFTR; Chloride channels; Extracellular nucleotides; P2Y receptors; Airway epithelium

Abbreviations: ABC, adenosine 5′-triphosphate-binding cassette; ADO, adenosine; ADP, adenosine 5′-diphosphate; AEC, airway epithelial cell; AMP, adenosine 5′-monophosphate; ASL, airway surface liquid; ATP, adenosine 5′-triphosphate; ATPase, adenosine triphosphatase; CaCC, Ca2+-activated chloride channel; cAMP,cyclic adenosine 5′-monophosphate; CF, cystic fibrosis; CFTR, cystic fibrosis transmembrane conductance regulator; CHO, Chinese hamster ovary; ClC, chloridechannels; DC, dendritic cell; ENaC, epithelial sodium channel; IDO, indoleamine 2,3-dioxygenase; mRNA, messenger ribonucleic acid; NBD, nucleotide-bindingdomain; NSAP, nonspecific alkaline phosphatase; ORCC, outwardly rectifying chloride channel; PCL, periciliary liquid; PKA, protein kinase A; PKC, protein kinaseC; RNA, ribonucleic acid; SLPI, secretory leukocyte protease inhibitor; UDP, uridine 5′-diphosphate; UTP, uridine 5′-triphosphate.

☆ The author⁎ CorrespondE-mail add

0163-7258/$ -doi:10.1016/j.p

Contents

1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7202. Does cystic fibrosis transmembrane conductance regulator control adenosine 5′-triphosphate

release? A controversial issue . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7203. Role of P2 receptors in the regulation of cystic fibrosis transmembrane conductance

regulator and other chloride channels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7223.1. Cystic fibrosis transmembrane conductance regulator and P2Y1 receptor. . . . . . . . . . . . . 7223.2. Cystic fibrosis transmembrane conductance regulator, chloride channels and P2Y2 receptors . . 7223.3. Cystic fibrosis transmembrane conductance regulator, P2Y4 and P2Y6 receptors . . . . . . . . . 7233.4. Cystic fibrosis transmembrane conductance regulator and P2X receptors. . . . . . . . . . . . . 724

s attest that there is no conflicts of interest.ing author.resses: [email protected], [email protected] (B. Marcet).

see front matter © 2006 Elsevier Inc. All rights reserved.harmthera.2006.05.010

Page 2: Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis

720 B. Marcet, J.-M. Boeynaems / Pharmacology & Therapeutics 112 (2006) 719–732

4. Physiological role of extracellular nucleotides in epithelia . . . . . . . . . . . . . . . . . . . . . . . . 7245. Extracellular nucleotides as therapeutic agents in cystic fibrosis . . . . . . . . . . . . . . . . . . . . . 7256. Airway inflammation in cystic fibrosis and its potential modulation by nucleotides . . . . . . . . . . . 7267. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 728Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 728References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 728

1. Introduction

This review provides an update on the current understandingof the relationship between cystic fibrosis transmembraneconductance regulator (CFTR) and extracellular nucleotidesand the use of nucleotides as therapeutic agents for treatingcystic fibrosis (CF) patients. We discuss published data,preliminary data and assumptions that concern the interactionbetween CFTR and extracellular nucleotides.

CF is one of the most common lethal autosomal recessivegenetic diseases caused by mutations in CFTR (Riordan et al.,1989), a cyclic adenosine 5′-monophosphate (cAMP)-regulatedchloride channel (ClC) localized in the apical membrane ofepithelial cells (Cheng et al., 1991). Two different hypothesesusing known properties of CFTR has emerged to attempt toexplain lung CF pathogenesis: the “low volume hypothesis” andthe “high salt hypothesis” (for review, see Wine, 1999).Interestingly, both hypotheses consider CFTR's role in saltabsorption to be primary. According to the classical view, called“low volume hypothesis” (Matsui et al., 1998; for review, seeWine, 1999), dysfunction in CFTR activity leads to a defect inCl− secretion and possibly a hyperabsorption of Na+ throughthe epithelial sodium channel (ENaC) (Knowles et al., 1991;Stutts et al., 1995; Mall et al., 2000; Kunzelmann &Mall, 2001)which ultimately results in severe damage of the airways andother exocrine organs, such as pancreas and intestine. In thelungs, the CFTR defect leads to a decrease in the airway surfaceliquid (ASL) covering the epithelia and thickened mucussecretions that severely impair mucociliary clearance, thuspromoting lung infections. According to this hypothesis, bothnormal and CF ASL have plasma-like levels of salt (Matsui etal., 1998). The other theory, the “high salt hypothesis” (Smith etal., 1996; Zabner et al., 1998), postulates that normal ASL haslow levels of salt as a result of salt absorption in excess of water.According to this hypothesis, lack or dysfunction of CFTR inCF causes reduced transepithelial Cl− conductance, salt ispoorly absorbed resulting in excessively salty ASL. The highsalt level in the ASL would interfere with the bacterial killing ofnatural antibiotics such as defensins (Goldman et al., 1997) andlysozyme.

In addition to functioning as a cAMP-activated Cl-channel,CFTR could also act as a regulator of other ion channels such asoutwardly rectifying chloride channels (ORCC) (Schwiebert etal., 1995; Julien et al., 1999), Ca2+-activated chloride channels(CaCC) (Wei et al., 1999) or ENaC (Stutts et al., 1995). Severalcontroversial studies have suggested that a CFTR-dependentrelease of cellular adenosine 5′-triphosphate (ATP) (Reisin etal., 1994; Schwiebert et al., 1995; Sugita et al., 1998; Urbach &

Harvey, 1999; Walsh et al., 2000) followed by the activation ofP2 nucleotide receptors could be involved in the regulation ofthese channels by CFTR. Such an effect suggests therefore arelationship between CFTR and P2 receptor signaling.

Extracellular nucleotides are important signaling moleculesthat mediate diverse biological effects, including ion transportregulation, by acting on nucleotide-gated ion channel P2X(1–7)

and G-protein coupled P2Y(1–14) receptors (for reviews, seeRalevic & Burnstock, 1998; Abbracchio et al., 2003; Leipziger,2003; Boeynaems et al., 2005). To date, 8 mammalian P2Yreceptors (P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, P2Y12, P2Y13,P2Y14) have been cloned and characterized (Lustig et al., 1993;Parr et al., 1994; Communi et al., 1995, 1997; Communi, Motteet al., 1996; Communi, Parmentier et al., 1996; Ralevic &Burnstock, 1998; Communi et al., 2001; Hollopeter et al., 2001;Abbracchio et al., 2003; Boeynaems et al., 2005). The P2Yreceptor family encompasses several subtypes: some areselective purinoceptors responsive to adenosine 5′-diphosphate(ADP: P2Y1, P2Y12, P2Y13) or ATP (P2Y11), others have aselectivity for uracil nucleotides, either uridine 5′-triphosphate(UTP: P2Y4) or uridine 5′-diphosphate (UDP: P2Y6), while theP2Y2 receptor has a mixed specificity and is activated to thesame extent by ATP and UTP. One ongoing area of researchsuggests that P2Y nucleotide receptors represent targetreceptors whose activation may have therapeutic value in CF.In particular, several studies emphasized the role of the P2Y2

receptor subtype, activated equipotently by ATP and UTP, as akey control for epithelial Cl− secretion by activating CaCC thatmay constitute a potential substitute to Cl− secretion defect inCF (Brown et al., 1991; Knowles et al., 1991; Cressman et al.,1999; Yerxa et al., 2002). According to the “low volumehypothesis”, aerosolized nucleotides have been evaluated forCF therapy as a means to increase ASL volume and to restoremucociliary clearance (see Section 5).

2. Does cystic fibrosis transmembraneconductance regulator control adenosine5′-triphosphate release? A controversial issue

CFTR is a member of the family of adenosine 5′-tripho-sphate-binding cassette (ABC) proteins that actively transport abroad range of substrates and include both eukaryotic andbacterial proteins, such as the multiple drug resistance protein(e.g., P-glycoprotein), the sulfonylurea receptor, the transporterfor antigen presentation, and the bacterial periplasmic per-meases (Riordan et al., 1989). CFTR contains 1480 amino acidsand consists of 2 homologous halves. Each half contains 6membrane-spanning segments and a nucleotide-binding domain

Page 3: Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis

721B. Marcet, J.-M. Boeynaems / Pharmacology & Therapeutics 112 (2006) 719–732

(NBD). The 2 halves of CFTR are linked by a cytoplasmicregulatory domain that contains a number of consensusphosphorylation sites for multiple protein kinases (Riordan etal., 1989). CFTR is unique among ABC proteins in that itstransmembrane domains comprise an ion channel that permitsbidirectional permeation of anions rather than vectorialtransport of solutes. CFTR has been well characterized as acAMP-regulated Cl-channel localized in apical membrane ofepithelial cells (Cheng et al., 1991).

P-glycoprotein, another member of the family of ABCtransporters, mediates the electrodiffusional movement of thenucleotide ATP (Abraham et al., 1993). Amino acid sequenceand structural similarities between CFTR and P-glycoproteinhave led Cantiello et al. to study the possibility that CFTR, inaddition to functioning as a cAMP-regulated Cl-channel, couldalso transport cellular ATP. They showed in a heterologousmodel that CFTR expression leads to protein kinase A (PKA)-induced ATP currents, inhibited by CFTR Cl-channel blockers.Their data argued in favor of ATP transport directly throughCFTR channels and suggested that CFTR is at the same time aCl-channel and an ATP channel (Reisin et al., 1994). Theobservations that P-glycoprotein and CFTR are associated withATP transport have also been supported by other studies usingpatch clamp and bulk ATP measurements in systems expressingCFTR (Pasyk & Foskett, 1997; Prat et al., 1996; Cantiello et al.,1998).

As illustrated in Fig. 1, once released in the extracellularmedium by a CFTR-dependent mechanism, ATP could controlother membrane channels or transporters (Schwiebert et al.,1995, 1999; Urbach & Harvey, 1999; Walsh et al., 2000;Kunzelmann & Mall, 2001), in particular ORCC. The nameORCC is related to electrophysiological properties (outwardlyrectifying) of certain Cl− conductances, but the molecularidentity of ORCC is still unknown (Gabriel et al., 1993;Schwiebert et al., 1994). A recent study has cloned andcharacterized ClC-3B, a novel alternative splicing variant ofClC-3 (ClC-3A), belonging to the CIC gene family of Cl-channels, and that is expressed predominantly in epithelial cells.The authors proposed that ClC-3B could be a candidate for the

Fig. 1. CFTR-dependent ATP release regulates other channels. CFTR that functions avia a distinct transporter or by regulating vesicle trafficking. Extracellular nucleoabsorption via ENaC by activating the P2Y2 receptor signaling pathway. Activation

ORCC molecule (Ogura et al., 2002). Schwiebert et al. (1995)showed that CFTR activation regulated ORCC by triggering thetransport of ATP out of the cell. A combination of a single-channel, whole cell recording and Ussing chamber short-circuitcurrent recordings revealed that ATP had to cross the membranefrom cytoplasm to extracellular space to exert its regulatoryeffect on Cl− conductances. In agreement with this assumption,the addition on the extracellular side of hexokinase or apyrase,enzymes which hydrolyze ATP, abolished the activation ofORCC, whereas the activity of CFTR was maintained(Schwiebert et al., 1995).

Although it seems now that there is an established relation-ship between CFTR and ATP transport in some particular cases,the problem is to determine whether ATP is released directlythrough the pore of CFTR or indirectly through anothermechanism controlled by CFTR (for reviews, see Devidas &Guggino, 1997; Schwiebert et al., 1999). One argument againstthe direct role of CFTR channel in the transport of ATP was thatthe diameter of the molecule of ATP (∼10 Å) would be largerthan that of the pore of CFTR estimated at approximately 5.3 Å(Illek et al., 1999). However, another study estimated that thediameter of CFTR pore could be as large as 10–13 Å (Linsdell& Hanrahan, 1998), enough to be permeant to ATP. Finally,other domains of the channel such as NBD1 could also take partin the passage of molecules through the membrane (Devidas &Guggino, 1997). Other studies argue in favor of ATP release,but through a distinct ATP channel controlled by CFTR. Studiescarried out in artificial membranes showed indeed that CFTRcannot by itself transport ATP (Li et al., 1996). Transientexpression of CFTR in Madin Darby canine kidney (MDCK)cells was associated with both Cl− and ATP currents uponCFTR activation by PKA (Sugita et al., 1998). The phos-phorylation of the regulatory domain, as well as hydrolysis ofATP by the NBD of CFTR, is necessary to detect an efflux ofATP. The authors concluded that the efflux of Cl− and ATP aredependent on CFTR but have a distinct sensitivity to blockers aswell as a distinct potential of reversion. That suggested aninteraction between CFTR and a transporter of ATP whosemolecular nature still remains unknown (Sugita et al., 1998).

s a cAMP-regulated Cl-channel, modulates the ATP release, directly or indirectlytides stimulate Ca2+-dependent Cl-channels (ORCC, CaCC) and inhibit Na+

of P2Y2 receptors may also modulate CFTR activity.

Page 4: Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis

722 B. Marcet, J.-M. Boeynaems / Pharmacology & Therapeutics 112 (2006) 719–732

Since CFTR possess a C-terminal motif known to interact withPDZ domain-containing proteins (Hall et al., 1998; Wang et al.,1998), it could be hypothesized that interaction between CFTRand putative ATP transporter might involve scaffolding proteinsand interaction with PDZ domain-containing proteins. Thedysfunction of CFTR could thus involve, in addition to thedirect defect of fluid and electrolytes transport, a defect of ATPsecretion outside the cell thus causing an additional defect ofother ion channels as well as other biological processes. Takentogether, these data suggest that CFTR does not transport ATPitself, but acts as a regulator of an associated transport systemfor ATP, a hypothesis also raised by al-Awqati (1995) andHiggins (1995).

Some other studies argue against a regulation of ATPtransport by the CFTR channel. These studies failed to detect anATP current or release of ATP dependent on CFTR withdifferent methodologies, like electrophysiological methods,radioactive ATP fluxes in reconstituted vesicles containingCFTR or intracellular calcium imaging in CFTR-expressingChinese hamster ovary (CHO) cells (Grygorczyk et al., 1996; Liet al., 1996; Reddy et al., 1996; Grygorczyk & Hanrahan, 1997;Marcet et al., 2003). An hypothesis then put forth is that therelease of ATP observed would be related to an artifact at thetime of the experiments of patch-clamp, like mechanicalstimulation of epithelial cells (Grygorczyk et al., 1996;Grygorczyk & Hanrahan, 1997; Watt et al., 1998). In asurprising way, in the same cellular model, CHO cells, someauthors detect a transport of ATP depending on the activation ofCFTR (Pasyk & Foskett, 1997; Urbach & Harvey, 1999)whereas others do not (Grygorczyk et al., 1996; Grygorczyk &Hanrahan, 1997; Marcet et al., 2003). Another explanation ofthese contradictory results obtained with the same CHO celllines would imply the presence of cofactors in the culturemedium, necessary to the transport of ATP (Schwiebert et al.,1999). Lastly, Boucher et al. did not find a difference in the levelof extracellular ATP in the airways of CF and non-CF subjects,suggesting that CFTR would not be implied in the transport ofATP in airways (Donaldson et al., 2000). To date, the role ofCFTR in ATP transport is not unambiguously demonstrated andis still debated in the literature.

3. Role of P2 receptors in theregulation of cystic fibrosis transmembraneconductance regulator and other chloride channels

Growing evidence suggests functional relationships betweenCFTR and P2Y as well as P2X receptors.

3.1. Cystic fibrosis transmembraneconductance regulator and P2Y1 receptor

Like the β2 adrenergic receptor, the P2Y1 receptor, whichis activated by ADP, and CFTR have, in their C-terminalend, a binding domain to PDZ domain-containing proteins.These proteins could act as scaffolds favoring an interactionbetween P2Y1 and CFTR in membrane microdomains (Hallet al., 1998; Naren et al., 2003; Fam et al., 2005). Recently,

we have shown that under particular conditions CFTR canmodulate the selectivity of P2Y1 receptor coupling to Gproteins (Marcet et al., 2004). We have shown that the P2Y1

receptor, which is known to couple primarily to Gq proteins,coupled to Gi/o proteins in the presence of CFTR. Thus, theexpression of CFTR in CHO-K1 cells induced a switch ofG-protein coupling selectivity of the P2Y1 receptor. Theseresults thus highlight the multipurpose role of CFTR channel(Marcet et al., 2004). In addition to this direct interactionwith CFTR, the P2Y1 receptor can modulate the activity ofboth CFTR and other Cl-channels. In Xenopus A6 cells,ADP, via P2Y1, increases both cAMP/PKA and Ca2+/proteinkinase C (PKC) activities and the PKC pathway is involvedin CFTR activation via potentiation of the PKA pathway(Guerra et al., 2004). The P2Y1 receptor is expressed in apicaland basolateral membranes of human airway epithelial cells(AEC) and its activation led to phospholipase C activation andCl− secretion, whose rate is regulated by the Ca2+-activated K+

channel (Son et al., 2004).

3.2. Cystic fibrosis transmembraneconductance regulator, chloride channels and P2Y2 receptors

In 1993–1994, the P2Y2 receptor, initially named P2U receptordue to its affinity for UTP, was cloned and characterized in mouseand human (Lustig et al., 1993, Parr et al., 1994). The P2Y2

receptor couples mainly to Gq and accessorily to Gi/o-proteins, isequipotently activated by ATP and UTP and triggers the Ca2+

signaling pathway (Ralevic & Burnstock, 1998). Earlier studieshad shown that ATP orUTP led to an increase in the Ca2+-activatedCl− secretion in normal and CF human AEC (Brown et al., 1991;Knowles et al., 1991; Mason et al., 1991). In situ hybridizationrevealed the expression of P2Y2 messenger ribonucleic acid(mRNA) in bronchial, bronchiolar and alveolar epithelia and insubmucosal gland epithelium (Yerxa et al., 2002). The study ofP2Y2-null mice has definitely demonstrated the exclusive role ofthat receptor in the stimulatory effect of ATP and UTP on trachealepithelial Cl− transport (Cressman et al., 1999). The molecularidentity of the Cl-channel involved remains to be elucidated. ATPand UTP have been shown to activate ORCC in human AEC (seeabove, Schwiebert et al., 1995) and rat tracheal epithelial cells(Hwang et al., 1996). As mentioned above, the molecular identityof ORCC remains unclear although it has been claimed that ClC-3B could be an ORCC (Ogura et al., 2002). However ORCC maynot be the only epithelial Cl-channel regulated by extracellularnucleotides. Indeed, nucleotides trigger an increase in intracellularCa2+ and may activate members of the CaCC family of Cl-channel(Mason et al., 1991;Knowles et al., 1991; Stutts et al., 1992).CaCCis the name given to Cl-channels that are activated by a rise inintracellular Ca2+. As for ORCC, themolecular identity of CaCC isstill unresolved. Recently, a new gene family of CI-channels(hClCA1–3) has been proposed to encode to CaCC (Gruber et al.,1998). Their expression may control the mucus production in non-CF and CFAEC (Toda et al., 2002; Hauber et al., 2004). However,these data remain very controversial since a study has shown thathClCA gene did not encode membrane proteins but rather solubleand secreted proteins (Gibson et al., 2005). Therefore, the

Page 5: Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis

723B. Marcet, J.-M. Boeynaems / Pharmacology & Therapeutics 112 (2006) 719–732

molecular identity of CaCC remains currently unresolved. Thus,nucleotides released in the extracellular environment may exerteffects on several distinct Ca2+-dependent Cl− conductances (e.g.,ORCC, CaCC, ClC, etc.).

In addition, it was recently shown that the P2Y2 receptorregulated also CFTR activity (Cantiello et al., 1994; Paradiso etal., 2001; Marcet et al., 2003). First, external ATP stimulatedhuman recombinant CFTR, transfected in C127i mouse mam-mary carcinoma cells, in a cAMP independent manner (Cantielloet al., 1994). Second, extracellular ATP orUTP could stimulate, inAEC, CFTR-dependent anion secretion through a Ca2+-indepen-dent PKC pathway subsequent to the activation of the Gq-coupledP2Y2 receptor (Paradiso et al., 2001). On the other hand, we haverecently shown an inhibitory effect of P2Y2 receptor on theactivity of CFTR channel in a heterologous model (Marcet et al.,2003). The activation of the P2Y2 receptor induced an inhibitionof the activity of CFTR by a mechanism independent of cAMP,but which could involve Ca2+ signaling, in the CFTR-expressingCHO-BQ1 cell line (Marcet et al., 2003). This action might berelated to the upregulation of CaCC which is observed in CFepithelia. Expression of CFTR has indeed been shown to inhibitCaCC-mediated current (Wei et al., 2001), while the loss of CFTRwas associated with CaCC upregulation in murine trachealepithelium (Tarran et al., 2002). Therefore it might be speculatedthat an inhibitory effect of UTP on CFTR could reinforce itsstimulatory effect on CaCC. Anyway this action should have noimpact on CF cells where CFTR is inactive.

The activation of P2Y2 receptors could also take part inthe inhibition of Na+ absorption via ENaC probably via thePKC pathway (Kunzelmann & Mall, 2001). As CF is alsocharacterized by hyperabsorption of Na+, some authors havedetermined the effect of mucosal UTP on transepithelial Na+

transport in primary cultures of human bronchial epithelia(Devor & Pilewski, 1999). They showed that UTP-inducedintracellular Ca2+ increase promoted a long-term inhibition oftransepithelial Na+ transport across normal and CF epithelialcells at least partly due to downregulation of a basolateralmembrane K+ conductance (Devor & Pilewski, 1999). Thus,UTP may have a dual therapeutic effect in CF airway bystimulating a Cl− secretory response and by inhibiting Na+

transport. However, the relative importance of an excessiveNa+ absorption in CF remains controversial (Joo et al.,2006).

All these data constitute the rationale of ongoing clinical trialsof aerosolized nucleotides in CF. In addition, because of itsexpression in the apical domain of the AEC, the P2Y2 receptorrepresents a potential target in the CF gene therapy, in order tosupport the internalisation of the vectors containing transgeneand thus to increase the efficacy of this therapy (Boucher, 1999;Kreda et al., 2000). The therapeutic role of nucleotides in CF islargely discussed in Section 5 of this review.

3.3. Cystic fibrosis transmembraneconductance regulator, P2Y4 and P2Y6 receptors

The concept that the action of nucleotides on the epithelialtransport of electrolytes and water involves multiple receptors

was confirmed by a gene targeting approach in mice. Indeed thenucleotide-stimulated Cl− secretion by the trachea was almostentirely abolished in P2Y2

−/− mice, whereas the nucleotide effectwas maintained completely in the jejunum and partially in thegallbladder (Cressman et al., 1999).

Following cloning of the murine P2Y4 gene (Suarez-Huertaet al., 2001), P2Y4-null mice were generated with aconventional gene targeting method. Sections of the mid-portion of the jejunum were mounted in Ussing chambers forbioelectric measurements. In wild-type animals, the apicalapplication of UTP increased the short circuit current. Thatresponse was abolished in P2Y4 knockout mice, whereas theyexhibited a normal response to the adenylyl cyclase activatorforskolin (Robaye et al., 2003). This work represents the firstclear-cut demonstration of a biological function of the P2Y4

receptor. It was thus demonstrated that the P2Y4 receptor isexpressed in murine jejunum and that its agonists are able tostimulate Cl− secretion in normal jejunum (Robaye et al.,2003). Similar observations were made in the colon (Ghanemet al., 2005). A response to nucleotides was also observed onthe basolateral side; that response was totally abolished in thecolon of P2Y4-null mice, whereas in the jejunum it waspartially decreased in both P2Y2- and P2Y4-deficient mice(Ghanem et al., 2005). Interestingly, the Cl− secretory responseto forskolin, mediated by CFTR, was potentiated by priorbasolateral addition of UTP and this potentiation wasabolished in P2Y4-null mice. Those data thus indicated thatthe P2Y4 receptor plays a dominant role in the gut, whereas inthe airways that role is played by the P2Y2 receptor.

In addition to the P2Y2 subtype, P2Y1, P2Y4, P2Y6, P2Y11

and P2Y12 mRNA could also be found in human airwaybronchial or nasal epithelial cells (Communi et al., 1999; Kim etal., 2004). Furthermore, there is pharmacological evidence thatin addition to the P2Y2 receptor, a functional receptorselectively responsive to UDP (presumably the P2Y6 subtype)is present on the apical surface of human nasal epithelial cells(Lazarowski et al., 1997). Recently, it was shown using Ussingchamber experiments that luminal UDP or the P2Y6 receptoragonist INS48823 induced both transient and persistentincreases in short circuit currents in the mouse trachea. Theauthors showed that activation of short circuit currents wasinhibited by the P2Y receptor blocker pyridoxalphosphate-6-azophenyl-2′,4′-disulfonic acid (PPADS). The transientresponse was inhibited by 4,4′-diisothiocyanatostilbène-2,2′-disulfonic acid (DIDS), whereas the persistent short circuitcurrent was inhibited by glibenclamide and by PKA blockerH-89. Moreover, sustained activation of short circuit currents byluminal UDP was inhibited by blocking basolateral K+ channelswith 293B. The authors suggested that activation of luminalP2Y6 receptors in the airways shifts electrolyte transporttowards secretion by increasing intracellular Ca2+ and activa-tion of PKA (Schreiber & Kunzelmann, 2005). Finally,Leipziger et al. have shown that basolateral P2Y6 receptors incolonic epithelial cells stimulated sustained NaCl secretion byway of a synergistic increase of intracellular Ca2+ and cAMP(Kottgen et al., 2003). In support of these data, the authorsdemonstrated that in Xenopus oocytes expressing the P2Y6

Page 6: Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis

724 B. Marcet, J.-M. Boeynaems / Pharmacology & Therapeutics 112 (2006) 719–732

receptor, UDP transiently activated the Ca2+-activated Cl−

current and subsequently CFTR (Kottgen et al., 2003).

3.4. Cystic fibrosis transmembraneconductance regulator and P2X receptors

In addition to activating P2Y receptor subtypes, extracellularATP could stimulate epithelial P2X receptors (Taylor et al.,1999; Zsembery et al., 2003). ATP-induced P2X receptoractivation promoted a sustained increase in intracellular Ca2+

concentration and potentiated Ca2+-dependant Cl− transportacross nasal mucosa of CF patients as well as across non-CFnasal mucosa. Thus, P2X receptors may provide additionaltargets for extracellular nucleotide therapy of CF (Taylor et al.,1999; Zsembery et al., 2003).

4. Physiological role of extracellular nucleotides in epithelia

The concept that adenine and uridine nucleotides function asextracellular signaling molecules has been markedly expandedin the last decade. The significance of nucleotides asextracellular molecules is also underscored by the ubiquitousdistribution of several classes of ectoenzymes that catalyzenucleotide breakdown and interconversion (Zimmermann,1996).

Normal airway epithelia are protected by a thin liquid layer:the ASL. ASL is divided into 2 layers: a mucus layer (height:∼7–70 μm in vivo) containing mucins and a periciliary liquid(PCL) layer (height: ∼7 μm), which keeps mucins at asufficient distance from the underlying epithelium to optimizecilia beating and mucus clearance (Knowles & Boucher, 2002;Tarran et al., 2006). Recent studies have demonstrated thecrucial role of external nucleotides in maintaining physiologicalASL volume (for review, see Tarran et al., 2006). IntracellularATP levels are∼5–10 mM, so relatively little cellular ATP mustbe released to reach a concentration large enough to activate P2receptors. Numerous studies showed that airway epitheliarelease ATP under basal conditions (Taylor et al., 1998;Donaldson et al., 2000) and in response to various mechanicalstimulations such as membrane stretch (Homolya et al., 2000),shear stress (Grygorczyk & Hanrahan, 1997; Watt et al., 1998;Tarran et al., 2005, 2006) and hypotonicity-induced swelling(Wang et al., 1996; Musante et al., 1999; Braunstein et al.,2001). Pathogen invasion is another mechanism that can induceATP release. Recently, it has been shown that flagellin ofPseudomonas aeruginosa inhibited ENaC by inducing ATPrelease from airway epithelia (Kunzelmann et al., 2006). ATP isalso released from tracheal epithelium by parainfluenza virus(Kunzelmann et al., 2004). UTP is co-released with ATP but inlower amounts (Lazarowski & Boucher, 2001). Lazarowski andHarden (1999) have previously described sensitive meth-odologies that allowed to measure UTP concentration. Theconcentration of UTP measured in the medium bathing ∼3·105

tissue culture cells under basal conditions ranged from 1 to5 nM and represented approximately 1/3 of the ATPconcentration. Since no difference in luminal UTP concentra-tion was found between normal and CF cells under either

resting or mechanically stimulated conditions (Lazarowski &Harden, 1999), it is also unlikely that CFTR was involved inUTP release by AEC. UDP-glucose is also released, by adistinct mechanism involving vesicle transport during traffick-ing of glycoproteins to the plasma membrane (Lazarowski et al.,2003).

Since under normal conditions, the mucus layer and the PCLare in continuous motion propelled by the ciliary beat cycle(∼40 μm/s), shear forces generated by the ASL movement mayprovide a mechanical stimulus that induces nucleotide release(Tarran et al., 2006). Moreover, bilateral release of UTP andATP may be triggered in some pathophysiological conditions inwhich the laminal PCL flow is disrupted by a mechanicalstimulus (e.g., during inhalation of foreign particles) or byincreased shear due to increased rates of airflow (e.g., cough).Although ATP and UTP concentrations measured in the dilutedmedium bathing the mucosal surface (∼200 μl/cm2) of primarycultures of resting human nasal epithelial cells seemed too lowto activate P2 receptors (Lazarowski & Harden, 1999),nucleotide levels in the thin (1 μl/cm2 or ∼20 μm of depth)film of ASL that covers the AEC surface grown at the air–liquidinterface (Jayaraman et al., 2001) might approach thresholdvalues for P2 receptor stimulation (0.1–1 μM). Furthermore theamounts released in response to mechanical stimulation mightbe sufficient to promote autocrine stimulation of P2Y receptorsas well as paracrine activation of nonepithelial P2Y and P2Xreceptors such as submucosal fibroblasts, smooth muscles andinflammatory cells (Lazarowski & Harden, 1999; Homolya etal., 2000; Tarran et al., 2006).

ATP can modulate the airway epithelium ion transport via theP2Y2 receptor but also via conversion into adenosine (ADO) byectonucleotidases. Recently, it has been shown that the ecto-adenosine triphosphatase (ATPase) activity in the mucosalsurface increased toward areas most susceptible to airwayobstruction (nose<bronchi≪bronchioles) (Picher et al., 2004).Boucher et al. have identified a nonspecific alkaline phospha-tase (NSAP) as the major ectonucleotidase in mucosal surfaceof airways. NSAP regulated nucleotide concentration in CF andwould be responsible for the termination of aerosolizednucleotide-mediated mucociliary clearance in the lungs of CFpatients (Picher et al., 2004). For example, they showed that themucosal epithelial surface eliminated P2 receptor agonists(ATP=UTP>ADP>UDP) at 3-fold higher rates than theserosal surface (Picher et al., 2004). Furthermore, the ecto-ATPase activity and mRNA expression of mucosally restrictedNSAP were 3-folds higher in bronchial epithelial cells from CFpatients than from healthy subjects, probably as a consequenceof chronic inflammation and infection. This indicates that in CFATP elimination and ADO accumulation on the mucosal surfaceare enhanced (Picher et al., 2004). Mucosal ADO levels reached2-fold higher values in CF epithelial cells. Boucher et al.(Lazarowski et al., 2004) have suggested that ATP but alsoADO could play a key role in regulating ASL volumehomeostasis (see Fig. 2). Indeed, basal ATP levels under restingconditions are insufficient to efficiently activate CaCC and tosustain normal ASL volume (Lazarowski et al., 2004). On theother hand, resting airway epithelia exhibit extracellular ADO

Page 7: Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis

Fig. 2. Extracellular nucleotides modulate the airway surface liquid volume and the mucociliary clearance. (A) Extracellular nucleotides regulate the rates of Na+

absorption and Cl− secretion to adjust PCL volume in non-CF airway epithelia. External ATP activates P2Y2 receptors that trigger Ca2+-dependent Cl− secretion via

CaCC and inhibit Na+ absorption via ENaC. ATP is degraded into ADO by NSAP and ADO activates A2B receptors which stimulate cAMP-dependent Cl− secretionvia CFTR. (B) In CF epithelia, the CFTR defect leads to PCL depletion as a result of a higher basal rate of Na+ absorption, the failure to inhibit Na+ transport rates andthe failure to initiate Cl− secretion via CFTR. In addition the upregulation of NSAP in response to chronic inflammation inhibits the P2Y2-mediated action ofnucleotides on other Cl− and Na+ channels.

725B. Marcet, J.-M. Boeynaems / Pharmacology & Therapeutics 112 (2006) 719–732

concentrations sufficient to activate CFTR (Huang et al., 2001).This activation may involve either A2A or A2B receptors, bothcoupled to cAMP pathway. The effect of ADO on ion transportis mediated by A2B receptors in the mouse trachea (Kornerup etal., 2005), human bronchial epithelial cells (Clancy et al., 1999)and human Calu-3 cells (Huang et al., 2001). The A2A receptoris also expressed by bronchial epithelial cells and seems to beinvolved in the remodeling of the airway epithelium (Allen-Gipson et al., 2006). A role for A1 receptors in airway epithelialion transport has also been proposed (Rugolo et al., 1993;Szkotak et al., 2001). Increased ADO is not expected to improvemucociliary clearance in CF lungs because A2B receptor-mediated stimulation of CFTR channel activity is defective(Boucher, 1994a, 1994b), although an A2B receptor-mediatedactivation of R117H-CFTR has been demonstrated (Clancy etal., 1999). Furthermore, ADOmight have a deleterious effect onNa+ hyperabsorption, since it has been demonstrated that anelevation of cAMP in the CF airway epithelium can enhanceNa+ absorption (Boucher et al., 1986). Since the ADO receptor-CFTR pathway is defective in CF patients, the ASL volumeseems rather dependent on mechanically stimulated ATP release(i.e., coughing, wheezing, and clapping) and CaCC activation(Picher et al., 2004), although that mechanism might becompromised by the upregulation of ectonucleotidases degrad-ing ATP and also UTP.

In addition to stimulating Cl− and water secretion, nucleo-tides can act on the other components of the mucociliaryescalator: mucin secretion and cilia beating. ATP stimulatescilia beating in human nasal epithelial cells via activation of theP2Y2 receptor and via degradation into ADO and activation ofA2B receptors (Wong & Yeates, 1992; Morse et al., 2001). Inhuman nasal epithelial cells, UTP and ATPγS, acted assecretagogues on mucin secretion via Ca2+-dependent pathwaysinvolving P2Y2 and possibly P2Y11 receptors (Chen et al.,

2001; Choi et al., 2003). In addition, UTP and ATP alsostimulate mucin secretion from epithelial goblet cells (Abdullahet al., 1996; Roger et al., 2000). Since CF is characterized byunderhydrated mucus and hypersecretion of mucins (Rose &Voynow, 2006; Voynow et al., 2006) that alter airwaymucociliary clearance, one wonders whether the P2Y-inducedmucin release may be beneficial in CF therapy in promotingmucociliary defense and clearance or rather might contribute topromote airway obstruction.

In summary, there is a complex extracellular nucleotidesignaling at the airway surface involving several ectonucleo-tidases that metabolized nucleotides (Zimmermann, 1996).Thus a large variety of nucleotides and nucleosides arepresent in ASL and may induce diverse and complex cellulareffects by acting on both ADO and nucleotide P2Y and P2Xreceptors.

5. Extracellular nucleotides astherapeutic agents in cystic fibrosis

In CF, defective cAMP-mediated regulation of Cl− con-ductance in AEC, as a result of CFTR mutations, limits thecapacity to secrete Cl−. As a result, CFTR dysfunctions lead to areduced ASL volume with the formation of viscous andunderhydrated mucus that obstructs the CF airways and favorsinfections in CF patients (Fig. 2). However, regulation of Cl−

conductance by other mechanisms, such as the CaCC via a risein intracellular Ca2+, is functional in CF epithelial cells,providing an opportunity to bypass the defective CFTR activity.P2Y nucleotide receptors, notably the P2Y2 subtype, are widelydistributed and have been shown to be prominently localized,like CFTR, to the apical membrane of AEC (Cressman et al.,1999, Homolya et al., 1999). These findings led to the proposalthat ATP or UTP may be used as therapeutic agents for lung

Page 8: Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis

726 B. Marcet, J.-M. Boeynaems / Pharmacology & Therapeutics 112 (2006) 719–732

disease in CF epithelium in counterbalancing both the Cl−

secretion defect by activating CaCC and the hyperabsorption ofNa+ by inhibiting ENaC (Knowles et al., 1991; Mason et al.,1991; Mall et al., 2000) and thereby promoting hydration ofairway secretions.

This observation has been made both in vitro, in humannasal epithelial cells, and in vivo: indeed superfusion ofnucleotides, combined to the ENaC blocker amiloride,decreased the nasal transepithelial potential difference bothin normal subjects and in CF patients, whose response hadactually a higher magnitude (Knowles et al., 1991).Furthermore, nucleotides enhance other activities of AECwhich are involved in the mucociliary clearance, such assubmucosal gland secretion and ciliary beating (Bennett et al.,1996). As UTP and ATP were equipotent, pyrimidinenucleotides seem preferable, since they are not degradedinto ADO which induces bronchoconstriction and possiblyairway inflammation (Blackburn et al., 1998; Chunn et al.,2001; for review, see Mohsenin & Blackburn, 2006). A PhaseI study in normal adults demonstrated that aerosolized UTP,alone or combined to amiloride, was safe and enhancedmucociliary clearance (Olivier et al., 1996). In adult CFpatients, aerosolized UTP and amiloride in combinationimproved mucociliary clearance from the peripheral airwaysto near normal values (Bennett et al., 1996). Rapid hydrolysisby ectonucleotidases severely restricts the effectiveness ofnucleotides. The safety of a long-lasting dinucleotide analogof UTP (Up4U, INS365) has been demonstrated in a Phase Istudy involving children and adults with CF (Noone et al.,1999; Kellerman, 2002; Kellerman et al., 2002). dCp4U(INS37217, denufosol tetrasodium), another dinucleotide withan even longer duration of action, has been developedrecently (Yerxa et al., 2002). In AEC or explants, denufosolincreased Cl− and water secretion, cilia beat frequency, andmucin release. The combined effect of these actions wasconfirmed in an animal model of tracheal mucus velocitywhich showed that a single administration of denufosolsignificantly enhanced mucus transport for at least 8 hr afterdosing (Yerxa et al., 2002).

The safety of denufosol was demonstrated in a Phase I/PhaseII trial that involved both adult and pediatric patients with CFwho were administered denufosol or placebo twice daily for5 days (Deterding et al., 2005). In this study, denufosol had noeffect on the lung function, as assessed by the 1-sec forcedexpiratory volume (FEV1), but increased significantly theexpectorated sputum. The results of Phase II clinical trials,funded by the CF Foundation, have been recently reported in apreliminary form. According to Inspire Pharmaceuticals,subjects receiving denufosol (10 mg, 20 mg, 40 mg and60 mg) for 4 weeks had significantly better lung function, asassessed by the FEV1 or forced expiratory flow 25–75%, thanpatients receiving placebo (http://www.inspirepharm.com).Inspire has planned a Phase III program to validate theuse of denufosol as an early intervention therapy for treat-ment of CF patients with mild lung disease (http://www.inspirepharm.com/pipeline.html). Orphan drug status has beengranted to denufosol by both the Food and Drug Adminis-

tration (FDA) and the European Medicines EvaluationAgency (EMEA).

6. Airway inflammation in cystic fibrosisand its potential modulation by nucleotides

Accumulated evidence suggests that early pulmonaryinflammation pathogenesis in CF may be associated with anabnormal increase in the production of pro-inflammatorycytokines in the CF lung, even in the absence of infectiousstimuli (Moss et al., 2000; Stecenko et al., 2001; Puchelle,2002). This dysregulation of cytokine generation by CFepithelial cells has been related with CFTR dysfunction(Moss et al., 1996). Indeed, in CF, production of pro-inflammatory cytokines like IL-1, IL-6, IL-8 or tumornecrosis factor-α was found upregulated whereas IL-10, apotent anti-inflammatory cytokine, was found downregulatedin peripheral blood monocytes as well as in airway cell lines(Bonfield et al., 1995; Tabary et al., 1998). Finally, it hasbeen suggested that a change from Th2 type immuneresponse, most frequent in CF, to a Th1 type immuneresponse might improve the prognosis in CF patients (Moseret al., 2000).

Over the past 15 years, several evidences showed that theimmune system can have profound effect on epithelial function(Berin et al., 1999). The dendritic cells (DC) are immune cellsinitially described in lymphoid organs and having a potentcapacity for presenting an antigen to lymphocytes. DC havebeen recently shown in the normal human lung at the level ofthe bronchioles. The number and state of differentiation of DCvary as a function of the epithelial microenvironment whichseems necessary in the differentiation of DC (Soler et al., 1991).In addition, a very recent study has shown cell–cell interactionbetween DC and epithelial cells (Lambrecht & Hammad, 2003).DC may have a significant role in the pulmonary immuneresponse where they play a pivotal role notably in the initiationof Th2 response-mediated lung inflammation (McWilliam et al.,1995; Masten & Lipscomb, 2000). On the other hand, AEC mayactively influence the activating properties of DC (Rimoldi etal., 2005).

In addition to the regulation of ion transport and othercomponents of mucociliary clearance, nucleotides could havepleiotropic effects on the inflammatory process in CF, both onepithelial cells and infiltrating immune cells. Little is knownabout the effects of nucleotides on the release of cytokines,chemokines, defensins and other proteins involved in theinflammatory reaction by AEC. We have recently investigatedthis question in detail by an approach combining microarrayexperiments, ELISA of specific proteins and chemotaxisexperiments. We show that nucleotides regulate CCL20 andIL-8 release, and thereby modulate immune cells recruitment(Marcet et al., submitted for publication). On the other hand ithas been reported that ATP and UTP promote the secretion ofthe secretory leukocyte protease inhibitor (SLPI) in humantracheal epithelial cells (Merten et al., 1993, 1996, 1998).SLPI is an elastase inhibitor which has anti-inflammatory andantimicrobial properties (Hiemstra, 2002). SLPI may have

Page 9: Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis

727B. Marcet, J.-M. Boeynaems / Pharmacology & Therapeutics 112 (2006) 719–732

protective effects, notably in CF airway, by decreasingelastase-dependent lung destruction, and promoting hostdefense against inflammation and infection (Hiemstra, 2002).

The modulation of immune cells by extracellular nucleotideshas been extensively studied in the last years. High concentra-tions of nucleotides, released during tissue injury from activatedplatelets or broken cells, could be important mediators ofinflammation. Recent data indicate that several nucleotide P2Yand P2X receptors are involved in the modulation of theimmune response and can exert pro- or anti-inflammatoryeffects (Fig. 3). Previous studies carried out in our laboratoryhave shown that ATP inhibited the activation of human CD4+ Tlymphocytes (Duhant et al., 2002). It also induced the

Fig. 3. Epithelia intimely interact with immune system by releasing nucleotides and iand inflammatory mediators (cytokines, chemokines) apically and basolaterally thimmature dendritic cells and a subpopulation of T lymphocytes and CCL17/CCLinflammation sites. (B) Extracellular nucleotides also regulate inflammatory propertiIL-10 and IDO production and inhibits release of MCP-1 and MIP-1α from DC. Ainduces chemotaxis and degranulation of neutrophils.

maturation of human DC and inhibited their production ofIL-12 and increased that of IL-10 via activation of the P2Y11

receptor (Schnurr et al., 2000; La Sala et al., 2001; Wilkin etal., 2001, 2002). Furthermore, it has been shown thatextracellular ATP strongly upregulated, in human DC, theexpression of thrombospondin-1 and indoleamine 2,3-dioxy-genase (IDO), 2 mediators involved in immune tolerance(Marteau et al., 2005). These data indicated that extracellularATP, previously considered as a danger signal, could also act as apotent regulator of mediators playing key roles in immunetolerance. In addition, we have recently shown that ATP stronglyreduced the capacity of DC to attract monocytes and immatureDC by inhibiting the release of 2 chemokines: macrophage

nflammatory mediators. (A) Inflamed and infected epithelia release nucleotidesat attract immune cells at inflammation sites. The chemokine CCL20 attracts22 attract B and T cells. The cytokine IL-8 strongly attracts neutrophils ates of immune cells. UDP induces IL-8 release by DC, ATP upregulates TSP-1,TP prevents release of IL-2, IL-5 IL-10 and IFN-γ from T cells. Finally, UTP

Page 10: Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis

728 B. Marcet, J.-M. Boeynaems / Pharmacology & Therapeutics 112 (2006) 719–732

inflammatory protein-1alpha (MIP-1α/CCL3) and monocytechemotactic protein-1 (MCP-1) (Horckmans et al., 2006). On theother hand, UTP was known to potentiate neutrophil attractionand degranulation (Verghese et al., 1996; Meshki et al., 2004).Finally, other studies have recently indicated that UDP, themetabolite product of UTP, promoted IL-8 release from DC(Idzko et al., 2004) and monocytes (Warny et al., 2001).

Thus, it could be hypothesized that nucleotides released oraerosolized in the airways could produce effects not only onAEC but also on immune cells such as neutrophils, DC ormacrophages present in inflammation sites. The contribution ofthese actions to the therapeutic efficacy of denufosol remains tobe evaluated.

7. Conclusion

Extracellular nucleotides are signaling molecules that playan important role in airway epithelial homeostasis by mod-ulating ion transport, ASL volume and mucociliary clearance.Based on that physiological role, nucleotides are developed astherapeutic agents for CF. Furthermore extracellular nucleotidesmodulate immune cells function and recruitment and theirrelease by AEC may play a key role in airway inflammation.

Acknowledgments

This work was supported by Research in Brussels, the Fondsde la Recherche Scientifique Médicale (FRSM), the Frenchassociation Vaincre La Mucoviscidose (VLM) and the KingBaudouin Foundation (Fonds Forton). We thank Dr. DidierCommuni for helpful discussion.

References

Abbracchio, M. P., Boeynaems, J. -M., Barnard, E. A., Boyer, J. L., Kennedy,C., Miras-Portugal, M. T., et al. (2003). Characterization of the UDP-glucosereceptor (re-named here the P2Y14 receptor) adds diversity to the P2Yreceptor family. Trends Pharmacol Sci 24, 52−55.

Abdullah, L. H., Davis, S. W., Burch, L., Yamauchi, M., Randell, S. H.,Nettesheim, P., et al. (1996). P2u purinoceptor regulation of mucin secretionin SPOC1 cells, a goblet cell line from the airways. Biochem J 316,943−951.

Abraham, E. H., Prat, A. G., Gerweck, L., Seneveratne, T., Arceci, R. J., Kramer,R., et al. (1993). The multidrug resistance (mdr1) gene product functions asan ATP channel. Proc Natl Acad Sci U S A 90(1), 312−316.

al-Awqati, Q. (1995). Regulation of ion channels by ABC transporters thatsecrete ATP. Science 269(5225), 805−806.

Allen-Gipson, D. S., Wong, J., Spurzem, J. R., Sisson, J. H., & Wyatt, T. A.(2006). Adenosine A2A receptors promote adenosine-stimulated woundhealing in bronchial epithelial cells. Am J Physiol Lung Cell Mol Physiol290(5), L849−L855.

Bennett, W. D., Olivier, K. N., Zeman, K. L., Hohneker, K. W., Boucher, R. C.,& Knowles, M. R. (1996). Effect of uridine 5′-triphosphate plus amilorideon mucociliary clearance in adult cystic fibrosis. Am J Respir Crit Care Med153, 1796−1801.

Berin, M. C., McKay, D. M., & Perdue, M. H. (1999). Immune-epithelialinteractions in host defense. Am J Trop Med Hyg 60, 16−25.

Blackburn, M. R., Datta, S. K., & Kellems, R. E. (1998). Adenosinedeaminase-deficient mice generated using a two-stage genetic engineeringstrategy exhibit a combined immunodeficiency. J Biol Chem 273(9),5093−5100.

Boeynaems, J. -M., Communi, D., Gonzalez, N. S., & Robaye, B. (2005).Overview of the P2 receptors. Semin Thromb Hemost 31(2), 139−149.

Bonfield, T. L., Konstan, M. W., Burfeind, P., Panuska, J. R., Hilliard, J. B., &Berger, M. (1995). Normal bronchial epithelial cells constitutively producethe anti-inflammatory cytokine interleukin-10, which is downregulated incystic fibrosis. Am J Respir Cell Mol Biol 13, 257−261.

Boucher, R. C. (1994a). Human airway ion transport: Part 1. Am J Respir CritCare Med 150, 271−281.

Boucher, R. C. (1994b). Human airway ion transport: Part 2. Am J Respir CritCare Med 150, 581−593.

Boucher, R. C. (1999). Status of gene therapy for cystic fibrosis lung disease.J Clin Invest 103, 441−445.

Boucher, R. C., Stutts, M. J., Knowles, M. R., Cantley, L., & Gatzy, J. T.(1986). Na+ transport in cystic fibrosis respiratory epithelia. Abnormalbasal rate and response to adenylate cyclase activation. J Clin Invest 78(5),1245−1252.

Braunstein, G. M., Roman, R. M., Clancy, J. P., Kudlow, B. A., Taylor, A. L.,Shylonsky, V. G., et al. (2001). Cystic fibrosis transmembrane conductanceregulator facilitates ATP release by stimulating a separate ATP releasechannel for autocrine control of cell volume regulation. J Biol Chem 276(9),6621−6630.

Brown, H. A., Lazarowski, E. R., Boucher, R. C., & Harden, T. K. (1991).Evidence that UTP and ATP regulate phospholipase C through a commonextracellular 5V-nucleotide receptor in human airway epithelial cells. MolPharmacol 40(5), 648−655.

Cantiello, H. F., Prat, A. G., Reisin, I. L., Ercole, L. B., Abraham, E. H., Amara,J. F., et al. (1994). External ATP and its analogs activate the cystic fibrosistransmembrane conductance regulator by a cyclic AMP-independentmechanism. J Biol Chem 269, 11224−11232.

Cantiello, H. F., Jackson, G. R., Jr., Grosman, C. F., Prat, A. G., Borkan, S. C.,Wang, Y., et al. (1998). Electrodiffusional ATP movement through the cysticfibrosis transmembrane conductance regulator. Am J Physiol 274(3 Pt 1),C799−C809.

Chen, Y., Zhao, Y. H., & Wu, R. (2001). Differential regulation of airway mucingene expression and mucin secretion by extracellular nucleotide tripho-sphates. Am J Respir Cell Mol Biol 25(4), 409−417.

Cheng, S. H., Rich, D. P., Marshall, J., Gregory, R. J., Welsh, M. J., & Smith,A. E. (1991). Phosphorylation of the R domain by cAMP-dependantprotein kinase regulates the CFTR chloride channel. Cell 66, 1027−1036.

Choi, J. Y., Namkung, W., Shin, J. H., & Yoon, J. H. (2003). Uridine-5′-triphosphate and adenosine triphosphate gamma S induce mucin secretionvia Ca2+-dependent pathways in human nasal epithelial cells. ActaOtolaryngol 123(9), 1080−1086.

Chunn, J. L., Young, H. W., Banerjee, S. K., Colasurdo, G. N., & Blackburn,M. R. (2001). Adenosine-dependent airway inflammation and hyperre-sponsiveness in partially adenosine deaminase-deficient mice. J Immunol167(8), 4676−4685.

Clancy, J. P., Ruiz, F. E., & Sorscher, E. J. (1999). Adenosine and its nucleotidesactivate wild-type and R117H CFTR through an A2B receptor-coupledpathway. Am J Physiol 276, C361−C369.

Communi, D., Pirotton, S., Parmentier, M., & Boeynaems, J. -M. (1995).Cloning and functional expression of a human uridine nucleotide receptor.J Biol Chem 270, 30849−30852.

Communi, D., Motte, S., Boeynaems, J. M., & Pirotton, S. (1996).Pharmacological characterization of the human P2Y4 receptor. Eur JPharmacol 317, 383−389.

Communi, D., Parmentier, M., & Boeynaems, J. -M. (1996). Cloning, functionalexpression and tissue distribution of the human P2Y6 receptor. BiochemBiophys Res Commun 222, 303−308.

Communi, D., Govaerts, C., Parmentier, M., & Boeynaems, J. -M. (1997).Cloning of a human purinergic P2Y receptor coupled to phospholipase Cand adenylyl cyclase. J Biol Chem 272, 31969−31973.

Communi, D., Paindavoine, P., Place, G. A., Parmentier, M., & Boeynaems,J. M. (1999). Expression of P2Y receptors in cell lines derived from thehuman lung. Br J Pharmacol 127, 562−568.

Communi, D., Suarez Gonzalez, N., Detheux, M., Brézillon, S., Lannoy, V.,Parmentier, M., et al. (2001). Identification of a novel human ADP receptorcoupled to Gi. J Biol Chem 276, 41479−41485.

Page 11: Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis

729B. Marcet, J.-M. Boeynaems / Pharmacology & Therapeutics 112 (2006) 719–732

Cressman, V. L., Lazarowski, E. R., Homolya, L., Boucher, R. C., Koller, B. H.,& Grubb, B. R. (1999). Effect of loss of P2Y2 receptor gene expression onnucleotide regulation of murine epithelial Cl− transport. J Biol Chem 274,26461−26468.

Deterding, R., Retsch-Bogart, G., Milgram, L., Gibson, R., Daines, C., Zeitlin,P. L., et al. (2005). Safety and tolerability of denufosol tetrasodiuminhalation solution, a novel P2Y2 receptor agonist: results of a phase 1/phase2 multicenter study in mild to moderate cystic fibrosis. Cystic FibrosisFoundation Therapeutics Development Network. Pediatr Pulmonol 39(4),339−348.

Devidas, S., & Guggino, W. B. (1997). CFTR: domains, structure, and function.J Bioenerg Biomembr 29, 443−451.

Devor, D. C., & Pilewski, J. M. (1999). UTP inhibits Na+ absorption in wild-type and DeltaF508 CFTR-expressing human bronchial epithelia. Am JPhysiol 276(4 Pt 1), C827−C837.

Donaldson, S. H., Lazarowski, E. R., Picher, M., Knowles, M. R., Stutts, M. J.,& Boucher, R. C. (2000). Basal nucleotide levels, release, and metabolism innormal and cystic fibrosis airways. Mol Med 6, 969−982.

Duhant, X., Schandene, L., Bruyns, C., Gonzalez, N. S., Goldman, M.,Boeynaems, J. -M., et al. (2002). Extracellular adenine nucleotidesinhibit the activation of human CD4+ T lymphocytes. J Immunol 169(1),15−21.

Fam, S. R., Paquet, M., Castleberry, A. M., Oller, H., Lee, C. J., Traynelis, S.F., et al. (2005). P2Y1 receptor signaling is controlled by interaction withthe PDZ scaffold NHERF-2. Proc Natl Acad Sci U S A 102(22),8042−8047.

Gabriel, S. E., Clarke, L. L., Boucher, R. C., & Stutts, M. J. (1993). CFTR andoutward rectifying chloride channels are distinct proteins with a regulatoryrelationship. Nature 363(6426), 263−268.

Ghanem, E., Robaye, B., Leal, T., Leipziger, J., Van Driessche, W., Beauwens,R., et al. (2005). The role of epithelial P2Y2 and P2Y4 receptors in theregulation of intestinal chloride secretion. Br J Pharmacol 146(3),364−369.

Gibson, A., Lewis, A. P., Affleck, K., Aitken, A. J., Meldrum, E., & Thompson,N. (2005). hCLCA1 and mCLCA3 are secreted non-integral membraneproteins and therefore are not ion channels. J Biol Chem 280(29),27205−27212.

Goldman, M. J., Anderson, G. M., Stolzenberg, E. D., Kari, U. P., Zasloff, M., &Wilson, J. M. (1997). Human beta-defensin-1 is a salt-sensitive antibiotic inlung that is inactivated in cystic fibrosis. Cell 88(4), 553−560.

Gruber, A. D., Elble, R. C., Ji, H. L., Schreur, K. D., Fuller, C. M., & Pauli, B. U.(1998). Genomic cloning,molecular characterization, and functional analysisof human CLCA1, the first human member of the family of Ca2+-activatedCl− channel proteins. Genomics 54(2), 200−214.

Grygorczyk, R., & Hanrahan, J. W. (1997). CFTR-independant ATP releasefrom epithelial cells triggered by mechanical stimuli. Am J Physiol 272,C1058−C1066.

Grygorczyk, R., Tabcharani, J. A., & Hanrahan, J. W. (1996). CFTR channelsexpressed in CHO cells do not have detectable ATP conductance. J Mem-brane Biol 151, 139−148.

Guerra, L., Favia, M., Fanelli, T., Calamita, G., Svetlo, M., Bagorda, A., et al.(2004). Stimulation of Xenopus P2Y1 receptor activates CFTR in A6 cells.Pflugers Arch 449(1), 66−75.

Hall, R. A., Ostedgaard, L. S., Premont, R. T., Blitzer, J. T., Rahman, N., Welsh,M. J., et al. (1998). A C-terminal motif found in the β2-adrenergic receptor,P2Y1 receptor and cystic fibrosis transmembrane conductance regulatordetermines binding to the Na+/H+ exchanger regulatory factor family of PDZproteins. Proc Natl Acad Sci U S A 95, 8496−8501.

Hauber, H. P., Tsicopoulos, A., Wallaert, B., Griffin, S., McElvaney, N. G.,Daigneault, P., et al. (2004). Expression of HCLCA1 in cystic fibrosis lungsis associated with mucus overproduction. Eur Respir J 23(6), 846−850.

Hiemstra, P. S. (2002). Novel roles of protease inhibitors in infection andinflammation. Biochem Soc Trans 30(2), 116−120.

Higgins, C. F. (1995). P-glycoprotein and cell volume-activated chloridechannels. J Bioenerg Biomembr 27(1), 63−70.

Hollopeter, G., Jantzen, H. -M., Vincent, D., Li, G., England, L., Ramakrishnan,V., et al. (2001). Identification of the platelet ADP receptor targeted byantithrombotic drugs. Nature 409, 202−207.

Homolya, L., Watt, W. C., Lazarowski, E. R., Koller, B. H., & Boucher, R. C.(1999). Nucleotide-regulated calcium signaling in lung fibroblasts andepithelial cells from normal and P2Y2 receptor (−/−) mice. J Biol Chem274, 26454−26460.

Homolya, L., Steinberg, T. H., & Boucher, R. C. (2000). Cell to cellcommunication in response to mechanical stress via bilateral release of ATPand UTP in polarized epithelia. J Cell Biol 150(6), 1349−1360.

Horckmans, M., Marcet, B., Marteau, F., Bulte, F., Maho, A., Parmentier, M., etal. (2006). Extracellular adenine nucleotides inhibit the release of majormonocyte recruiters by human monocyte-derived dendritic cells. FEBS Lett580(3), 747−754.

Huang, P., Lazarowski, E. R., Tarran, R., Milgram, S. L., Boucher, R. C., &Stutts, M. J. (2001). Compartmentalized autocrine signaling to cysticfibrosis transmembrane conductance regulator at the apical membrane ofairway epithelial cells. Proc Natl Acad Sci U S A 98(24), 14120−14125.

Hwang, T. H., Schwiebert, E. M., & Guggino, W. B. (1996). Apical andbasolateral ATP stimulates tracheal epithelial chloride secretion via multiplepurinergic receptors. Am J Physiol 270(6 Pt 1), C1611−C1623.

Idzko, M., Panther, E., Sorichter, S., Herouy, Y., Berod, L., Geissler, M., et al.(2004). Characterization of the biological activities of uridine diphosphate inhuman dendritic cells: influence on chemotaxis and CXCL8 release. J CellPhysiol 201(2), 286−293.

Illek, B., Tam, A. W., Fischer, H., & Machen, T. E. (1999). Anion selectivity ofapical membrane conductance of Calu 3 human airway epithelium. PflugersArch 437(6), 812−822.

Jayaraman, S., Song, Y., Vetrivel, L., Shankar, L., & Verkman, A. S. (2001).Noninvasive in vivo fluorescence measurement of airway-surface liquiddepth, salt concentration, and pH. J Clin Invest 107(3), 317−324.

Joo, N. S., Irokawa, T., Robbins, R. C., & Wine, J. J. (2006). Hyposecretion, nothyperabsorption, is the basic defect of cystic fibrosis airway glands. J BiolChem 281(11), 7392−7398.

Julien, M., Verrier, B., Cerutti, M., Chappe, V., Gola, M., Devauchelle, G., et al.(1999). Cystic Fibrosis Transmembrane Conductance Regulator (CFTR)confers glibenclamide-sensitivity to outwardly rectifying chloride channel(ORCC) in Hi-5 insect cells. J Membr Biol 168, 229−239.

Kellerman, D. J. (2002). P2Y(2) receptor agonists: a new class of medicationtargeted at improved mucociliary clearance. Chest 121, 201S−205S.

Kellerman, D., Evans, R., Mathews, D., & Shaffer, C. (2002). Inhaled P2Y2receptor agonists as a treatment for patients with Cystic Fibrosis lungdisease. Adv Drug Deliv Rev 54, 1463−1474.

Kim, C. H., Kim, S. S., Choi, J. Y., Shin, J. H., Kim, J. Y., Namkung, W., et al.(2004). Membrane-specific expression of functional purinergic receptors innormal human nasal epithelial cells. Am J Physiol Lung Cell Mol Physiol287(4), L835−L842.

Knowles, M. R., & Boucher, R. C. (2002). Mucus clearance as a primary innatedefense mechanism for mammalian airways. J Clin Invest 109, 571−577.

Knowles, M. R., Clarke, L. L., & Boucher, R. C. (1991). Activation byextracellular nucleotides of chloride secretion in the airway epithelia ofpatients with cystic fibrosis. N Engl J Med 325, 533−538.

Kornerup, K. N., Page, C. P., & Moffatt, J. D. (2005). Pharmacologicalcharacterisation of the adenosine receptor mediating increased ion transportin the mouse isolated trachea and the effect of allergen challenge. Br JPharmacol 144(7), 1011−1016.

Kottgen, M., Loffler, T., Jacobi, C., Nitschke, R., Pavenstadt, H., Schreiber,R., et al. (2003). P2Y6 receptor mediates colonic NaCl secretion viadifferential activation of cAMP-mediated transport. J Clin Invest 111(3),371−379.

Kreda, S. M., Pickles, R. J., Lazarowski, E. R., & Boucher, R. C. (2000). G-protein-coupled receptors as targets for gene transfer vectors using naturalsmall-molecule ligands. Nat Biotechnol 18(6), 635−640.

Kunzelmann, K., & Mall, M. (2001). Pharmacotherapy of the ion transportdefect in cystic fibrosis. Clin Exp Pharmacol Physiol 28, 857−867.

Kunzelmann, K., Konig, J., Sun, J., Markovich, D., King, N. J., Karupiah, G.,et al. (2004). Acute effects of parainfluenza virus on epithelial electrolytetransport. J Biol Chem 279(47), 48760−48766.

Kunzelmann, K., Scheidt, K., Scharf, B., Ousingsawat, J., Schreiber, R.,Wainwright, B., et al. (2006). Flagellin of Pseudomonas aeruginosa inhibitsNa+ transport in airway epithelia. FASEB J 20(3), 545−546.

Page 12: Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis

730 B. Marcet, J.-M. Boeynaems / Pharmacology & Therapeutics 112 (2006) 719–732

Lambrecht, B. N., & Hammad, H. (2003). The other cells in asthma: dendriticcell and epithelial cell croostalk. Curr Opin Pulm Med 9, 34−41.

la Sala, A., Ferrari, D., Corinti, S., Cavani, A., Di Virgilio, F., & Girolomoni, G.(2001). Extracellular ATP induces a distorted maturation of dendritic cellsand inhibits their capacity to initiate Th1 responses. J Immunol 166(3),1611−1617.

Lazarowski, E. R., Paradiso, A. M., Watt, W. C., Harden, T. K., & Boucher, R. C.(1997). UDP activates a mucosal-restricted receptor on human nasalepithelial cells that is distinct from the P2Y2 receptor. Proc Natl Acad SciU S A 94(6), 2599−2603.

Lazarowski, E. R., & Boucher, R. C. (2001). UTP as an extracellular signalingmolecule. News Physiol Sci 16, 1−5.

Lazarowski, E. R., & Harden, T. K. (1999). Quantitation of extracellular UTPusing a sensitive enzymatic assay. Br J Pharmacol 127, 1272−1278.

Lazarowski, E. R., Shea, D. A., Boucher, R. C., & Harden, T. K. (2003). Releaseof cellular UDP-glucose as a potential extracellular signaling molecule.MolPharmacol 63, 1190−1197.

Lazarowski, E. R., Tarran, R., Grubb, B. R., van Heusden, C. A., Okada, S., &Boucher, R. C. (2004). Nucleotide release provides a mechanism for airwaysurface liquid homeostasis. J Biol Chem 279(35), 36855−36864.

Leipziger, J. (2003). Control of epithelial transport via luminal P2 receptors. AmJ Physiol Renal Physiol 284, F419−F432.

Li, C., Ramjeesingh, M., & Bear, C. E. (1996). Purified cystic fibrosistransmembrane conductance regulator (CFTR) does not function as an ATPchannel. J Biol Chem 271, 11623−11626.

Linsdell, P., & Hanrahan, J. W. (1998). Adenosine triphosphate-dependentasymmetry of anion permeation in the cystic fibrosis transmembraneconductance regulator chloride channel. J Gen Physiol 111, 601−614.

Lustig, K. D., Shiau, A. K., Brake, A. J., & Julius, D. (1993). Expressioncloning of an ATP receptor from mouse neuroblastoma cells. Proc NatlAcad Sci U S A 90, 5113−5117.

Mall, M., Wissner, A., Gonska, T., Calenborn, D., Kuehr, J., Brandis, M., et al.(2000). Inhibition of amiloride-sensitive epithelial Na+ absorption byextracellular nucleotides in human normal and cystic fibrosis airways. AmJ Respir Cell Mol Biol 23, 755−761.

Marcet, B., Chappe, V., Delmas, P., Gola, M., & Verrier, B. (2003). Negativeregulation of CFTR activity by extracellular ATP involves P2Y2 receptors inCFTR-expressing CHO cells. J Membr Biol 194, 21−32.

Marcet, B., Chappe, V., Delmas, P., & Verrier, B. (2004). Pharmacological andsignaling properties of endogenous P2Y1 receptors in cystic fibrosistransmembrane conductance regulator-expressing Chinese hamster ovarycells. J Pharmacol Exp Ther 309(2), 533−539.

Marteau, F., Gonzalez, N. S., Communi, D., Goldman, M., Boeynaems, J. -M.,& Communi, D. (2005). Thrombospondin-1 and indoleamine 2,3-dioxy-genase are major targets of extracellular ATP in human dendritic cells. Blood106(12), 3860−3866.

Mason, S. J., Paradiso, A. M., & Boucher, R. C. (1991). Regulation oftransepithelial ion transport and intracellular calcium by extracellular ATP inhuman normal and cystic fibrosis airway epithelium. Br J Pharmacol 103,1649−1656.

Masten, B. J., & Lipscomb, M. F. (2000). Dendritic cells: pulmonary immuneregulation and asthma. Monaldi Arch Chest Dis 55, 225−230.

Matsui, H., Grubb, B. R., Tarran, R., Randell, S. H., Gatzy, J. T., Davis, C. W., etal. (1998). Evidence for periciliary liquid layer depletion, not abnormal ioncomposition, in the pathogenesis of cystic fibrosis airways disease. Cell 95(7), 1005−1015.

McWilliam, A. S., Nelson, D. J., & Holt, P. G. (1995). The biology of airwaydendritic cells. Immunol Cell Biol 73(5), 405−413.

Merten, M. D., Breittmayer, J. P., Figarella, C., & Frelin, C. (1993). ATP andUTP increase secretion of bronchial inhibitor by human tracheal gland cellsin culture. Am J Physiol 265(5 Pt 1), L479−L484.

Merten, M. D., Kammouni, W., Renaud, W., Birg, F., Mattei, M. G., & Figarella,C. (1996). A transformed human tracheal gland cell line, MM-39, thatretains serous secretory functions. Am J Respir Cell Mol Biol 15(4),520−528.

Merten, M. D., Saleh, A., Kammouni, W., Marchand, S., & Figarella, C. (1998).Characterization of two distinct P2Y receptors in human tracheal gland cells.Eur J Biochem 251(1-2), 19−24.

Meshki, J., Tuluc, F., Bredetean, O., Ding, Z., & Kunapuli, S. P. (2004).Molecular mechanism of nucleotide-induced primary granule release inhuman neutrophils: role for the P2Y2 receptor. Am J Physiol Cell Physiol286(2), C264−C271.

Mohsenin, A., & Blackburn, M. R. (2006). Adenosine signaling in asthma andchronic obstructive pulmonary disease. Curr Opin Pulm Med 12(1),54−59.

Morse, D. M., Smullen, J. L., & Davis, C. W. (2001). Differential effects of UTP,ATP, and adenosine on ciliary activity of human nasal epithelial cells. Am JPhysiol Cell Physiol 280(6), C1485−C1497.

Moser, C., Kjaergaard, S., Pressler, T., Kharazmi, A., Koch, C., & Hoiby, N.(2000). The immune response to chronic Pseudomonas aeruginosa lunginfection in cystic fibrosis patients is predominantly of the Th2 type. APMIS108, 329−335.

Moss, R. B., Bocian, R. C., Hsu, Y. P., Dong, Y. J., Kemna, M., Wei, T., et al.(1996). Reduced IL-10 secretion by CD4+ T lymphocytes expressingmutant cystic fibrosis transmembrane conductance regulator (CFTR). ClinExp Immunol 106, 374−388.

Moss, R. B., Hsu, Y. P., & Olds, L. (2000). Cytokine dysregulation in activatedcystic fibrosis (CF) peripheral lymphocytes.Clin Exp Immunol 120, 252−518.

Musante, L., Zegarra-Moran, O., Montaldo, P. G., Ponzoni, M., & Galietta, L. J.(1999). Autocrine regulation of volume-sensitive anion channels in airwayepithelial cells by adenosine. J Biol Chem 274(17), 11701−11707.

Naren, A. P., Cobb, B., Li, C., Roy, K., Nelson, D., Heda, G. D., et al. (2003). Amacromolecular complex of beta 2 adrenergic receptor, CFTR, and ezrin/radixin/moesin-binding phosphoprotein 50 is regulated by PKA. Proc NatlAcad Sci U S A 100(1), 342−346.

Noone, P. G., Bennett, W. D., Regnis, J. A., Zeman, K. L., Carson, J. L., King,M., et al. (1999). Effect of aerosolized uridine-5′-triphosphate on airwayclearance with cough in patients with primary ciliary dyskinesia. Am JRespir Crit Care Med 160, 144−149.

Ogura, T., Furukawa, T., Toyozaki, T., Yamada, K., Zheng, Y. J., Katayama, Y.,et al. (2002). ClC-3B, a novel ClC-3 splicing variant that interacts withEBP50 and facilitates expression of CFTR-regulated ORCC. FASEB J 16(8), 863−865.

Olivier, K. N., Bennett, W. D., Hohneker, K. W., Zeman, K. L., Edwards, L. J.,Boucher, R. C., et al. (1996). Acute safety and effects on mucociliaryclearance of aerosolized uridine 5′-triphosphate +/− amiloride in normalhuman adults. Am J Respir Crit Care Med 154, 217−223.

Paradiso, A. M., Ribeiro, C. M. P., & Boucher, R. C. (2001). Polarized signalingvia purinoceptors in normal and cystic fibrosis airway epithelia. J GenPhysiol 117, 53−67.

Parr, C. E., Sullivan, D. M., Paradiso, A. M., Lazarowski, E. R., Burch, L. H.,Olsen, J. C., et al. (1994). Cloning and expression of a human P2Unucleotide receptor, a target for cystic fibrosis pharmacotherapy. Proc NatlAcad Sci U S A 91(8), 3275−3279.

Pasyk, E. A., & Foskett, J. K. (1997). Cystic fibrosis transmembrane conductanceregulator-associated ATP and adenosine 3′-phosphate 5′-phosphosulfatechannels in endoplasmic reticulum and plasma membranes. J Biol Chem 272(12), 7746−7751.

Picher, M., Burch, L. H., & Boucher, R. C. (2004). Metabolism of P2 receptoragonists in human airways: implications for mucociliary clearance andcystic fibrosis. J Biol Chem 279(19), 20234−20241.

Prat, A. G., Reisin, I. L., Ausiello, D. A., & Cantiello, H. F. (1996). Cellular ATPrelease by the cystic fibrosis transmembrane conductance regulator. Am JPhysiol 270(2 Pt 1), C538−C545.

Puchelle, E. (2002). Early bronchial inflammation in cystic fibrosis. J Soc Biol196, 29−35.

Ralevic, V., & Burnstock, G. (1998). Receptors for purines and pyrimidines.Pharmacol Rev 50(3), 413−492.

Reddy, M. M., Quinton, P. M., Haws, C., Wine, J. J., Grygorczyk, R.,Tabcharani, J. A., et al. (1996). Failure of the cystic fibrosis transmembraneconductance regulator to conduct ATP. Science 271(5257), 1876−1879.

Reisin, I. L., Prat, A. G., Abraham, E. H., Amara, J. F., Gregory, R. J., Ausiello,D. A., et al. (1994). The cystic fibrosis transmembrane conductance regulatoris a dual ATP and chloride channel. J Biol Chem 269, 20584−20591.

Rimoldi, M., Chieppa, M., Larghi, P., Vulcano, M., Allavena, P., & Rescigno,M. (2005). Monocyte-derived dendritic cells activated by bacteria or by

Page 13: Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis

731B. Marcet, J.-M. Boeynaems / Pharmacology & Therapeutics 112 (2006) 719–732

bacteria-stimulated epithelial cells are functionally different. Blood 106(8),2818−2826.

Riordan, J. R., Rommens, J. M., Kerem, B. -S., Alon, N., Rozmahel, R.,Grzelczak, Z., et al. (1989). Identification of the cystic fibrosis gene:cloning and characterization of complementary DNA. Science 245,1066−1072.

Robaye, B., Ghanem, E., Wilkin, F., Fokan, D., Van Driessche, W., Schurmans,S., et al. (2003). Loss of nucleotide regulation of epithelial chloride transportin the jejunum of P2Y4-null mice. Mol Pharmacol 63, 777−783.

Roger, P., Gascard, J. P., Bara, J., de Montpreville, V. T., Yeadon, M., & Brink,C. (2000). ATP induced MUC5AC release from human airways in vitro.Mediators Inflamm 9(6), 277−284.

Rose, M. C., & Voynow, J. A. (2006). Respiratory tract mucin genes and mucinglycoproteins in health and disease. Physiol Rev 86(1), 245−278.

Rugolo, M., Mastrocola, T., Whorle, C., Rasola, A., Gruenert, D. C., Romeo, G.,et al. (1993). ATP and A1 adenosine receptor agonists mobilize intracellularcalcium and activate K+ and Cl− currents in normal and cystic fibrosisairway epithelial cells. J Biol Chem 268(33), 24779−24784.

Schnurr, M., Then, F., Galambos, P., Scholz, C., Siegmund, B., Endres, S., et al.(2000). Extracellular ATP and TNF-alpha synergize in the activation andmaturation of human dendritic cells. J Immunol 165(8), 4704−4709.

Schreiber, R., & Kunzelmann, K. (2005). Purinergic P2Y6 receptors induce Ca2+ and CFTR dependent Cl− secretion in mouse trachea. Cell PhysiolBiochem 16(1-3), 99−108.

Schwiebert, E. M., Flotte, T., Cutting, G. R., & Guggino, W. B. (1994). BothCFTR and outwardly rectifying chloride channels contribute to cAMP-stimulated whole cell chloride currents. Am J Physiol 266, C1464−C1477.

Schwiebert, E. M., Egan, M. E., Hwang, T. H., Fulmer, S., Allen, S. S., Cutting,G. R., et al. (1995). CFTR regulates outwardly rectifying chloride channelsthrough an autocrine mechanism involving ATP. Cell 81, 1063−1073.

Schwiebert, E. M., Benos, D. J., Egan, M. E., Stutts, M. J., & Guggino, W. B.(1999). CFTR is a conductance regulator as well as a chloride channel.Physiol Rev 79(1 Suppl), S145−S166.

Smith, J. J., Travis, S. M., Greenberg, E. P., & Welsh, M. J. (1996). Cysticfibrosis airway epithelia fail to kill bacteria because of abnormal airwaysurface fluid. Cell 85(2), 229−236.

Soler, P., Tazi, A., Basset, F., & Hance, A. J. (1991). Pulmonary dendritic cells.Rev Mal Respir 8, 191−196.

Son, M., Ito, Y., Sato, S., Ishikawa, T., Kondo, M., Nakayama, S., et al. (2004).Apical and basolateral ATP-induced anion secretion in polarized humanairway epithelia. Am J Respir Cell Mol Biol 30(3), 411−419.

Stecenko, A. A., King, G., Torii, K., Breyer, R. M., Dworski, R., Blackwell, T.S., et al. (2001). Dysregulated cytokine production in human cystic fibrosisbronchial epithelial cells. Inflammation 25, 145−155.

Stutts,M. J., Chinet, T. C.,Mason, S. J., Fullton, J.M., Clarke, L. L.,&Boucher, R.C.(1992). Regulation of Cl− channels in normal and cystic fibrosis airway epithelialcells by extracellular ATP. Proc Natl Acad Sci U S A 89(5), 1621−1625.

Stutts, M. J., Canessa, C. M., Olsen, J. C., Hamrick, M., Cohn, J. A., Rossier, B.C., et al. (1995). CFTR as a cAMP-dependent regulator of sodium channels.Science 269, 847−850.

Suarez-Huerta, N., Pouillon, V., Boeynaems, J. -M., & Robaye, B. (2001).Molecular cloning and characterization of the mouse P2Y(4) nucleotidereceptor. Eur J Pharmacol 416(3), 197−202.

Sugita, M., Yue, Y., & Foskett, J. K. (1998). CFTR Cl− channel and CFTR-associated ATP channel: distinct pores regulated by common gates. EMBO J17, 898−908.

Szkotak, A. J., Ng, A. M., Sawicka, J., Baldwin, S. A., Man, S. F., Cass, C. E., etal. (2001). Regulation of K(+) current in human airway epithelial cells byexogenous and autocrine adenosine. Am J Physiol Cell Physiol 281(6),C1991−C2002.

Tabary, O., Zahm, J. M., Hinnrasky, J., Couetil, J. P., Cornillet, P., Guenounou,M., et al. (1998). Selective up-regulation of chemokine IL-8 expression incystic fibrosis bronchial gland cells in vivo and in vitro. Am J Pathol 153,921−930.

Tarran, R., Loewen, M. E., Paradiso, A. M., Olsen, J. C., Gray, M. A.,Argent, B. E., et al. (2002). Regulation of murine airway surface liquidvolume by CFTR and Ca2+-activated Cl− conductances. J Gen Physiol120, 407−418.

Tarran, R., Button, B., Picher, M., Paradiso, A. M., Ribeiro, C. M., Lazarowski,E. R., et al. (2005). Normal and cystic fibrosis airway surface liquidhomeostasis. The effects of phasic shear stress and viral infections. J BiolChem 280(42), 35751−35759.

Tarran, R., Button, B., & Boucher, R. C. (2006). Regulation of normal and cysticfibrosis airway surface liquid volume by phasic shear stress. Annu RevPhysiol 68, 543−561.

Taylor, A. L., Kudlow, B. A., Marrs, K. L., Gruenert, D. C., Guggino, W. B.,& Schwiebert, E. M. (1998). Bioluminescence detection of ATP releasemechanisms in epithelia. Am J Physiol 275(5 Pt 1), C1391−C1406.

Taylor, A. L., Schwiebert, L. M., Smith, J. J., King, C., Jones, J. R., Sorscher, E.J., et al. (1999). Epithelial P2X purinergic receptor channel expression andfunction. J Clin Invest 104(7), 875−884.

Toda, M., Tulic, M. K., Levitt, R. C., & Hamid, Q. (2002). A calcium-activatedchloride channel (HCLCA1) is strongly related to IL-9 expression andmucus production in bronchial epithelium of patients with asthma. J AllergyClin Immunol 109(2), 246−250.

Urbach, V., & Harvey, B. J. (1999). Regulation of intracellular Ca2+ by CFTR inChinese Hamster Ovary Cells. J Membr Biol 171, 255−265.

Verghese, M. W., Kneisler, T. B., & Boucheron, J. A. (1996). P2U agonistsinduce chemotaxis and actin polymerization in human neutrophils anddifferentiated HL60 cells. J Biol Chem 271(26), 15597−15601.

Voynow, J. A., Gendler, S. J., & Rose, M. C. (2006). Regulation of mucin genesin chronic inflammatory airway diseases. Am J Respir Cell Mol Biol 34(6),661−665.

Walsh, D. E., Harvey, B. J., & Urbach, V. (2000). CFTR regulation ofintracellular calcium in normal and cystic fibrosis human airway epithelia.J Membr Biol 177, 209−219.

Wang, Y., Roman, R., Lidofsky, S. D., & Fitz, J. G. (1996). Autocrine signalingthrough ATP release represents a novel mechanism for cell volumeregulation. Proc Natl Acad Sci U S A 93(21), 12020−12025.

Wang, S., Raab, R. W., Schatz, P. J., Guggino, W. B., & Li, M. (1998). Peptidebinding consensus of the NHE-RF-PDZ1 domain matches the C-terminalsequence of cystic fibrosis transmembrane conductance regulator (CFTR).FEBS Lett 427(1), 103−108.

Warny, M., Aboudola, S., Robson, S. C., Sevigny, J., Communi, D., Soltoff, S.P., et al. (2001). P2Y(6) nucleotide receptor mediates monocyte interleukin-8 production in response to UDP or lipopolysaccharide. J Biol Chem 276(28), 26051−26056.

Watt, W. C., Lazarowski, E. R., & Boucher, R. C. (1998). Cystic fibrosistransmembrane regulator-independent release of ATP. Its implications forthe regulation of P2Y2 receptors in airway epithelia. J Biol Chem 273(22),14053−14058.

Wei, L., Vankeerberghen, A., Cuppens, H., Eggermont, J., Cassiman, J. J.,Droogmans, G., et al. (1999). Interaction between calcium-activatedchloride channels and the cystic fibrosis transmembrane conductanceregulator. Pflügers Arch 438, 635−641.

Wei, L., Vankeerberghen, A., Cuppens, H., Cassiman, J. J., Droogmans, G., &Nilius, B. (2001). The C-terminal part of the R-domain, but not the PDZbinding motif, of CFTR is involved in interaction with Ca(2+)-activated Cl−

channels. Pflugers Arch 442, 280−285.Wilkin, F., Duhant, X., Bruyns, C., Suarez-Huerta, N., Boeynaems, J. -M., &

Robaye, B. (2001). The P2Y11 receptor mediates the ATP-induced maturationof human monocyte-derived dendritic cells. J Immunol 166(12), 7172−7177.

Wilkin, F., Stordeur, P., Goldman, M., Boeynaems, J. -M., & Robaye, B. (2002).Extracellular adenine nucleotides modulate cytokine production by humanmonocyte-derived dendritic cells: dual effect on IL-12 and stimulation of IL-10. Eur J Immunol 32(9), 2409−2417.

Wine, J. J. (1999). The genesis of cystic fibrosis lung disease. J Clin Invest 103(3), 309−312.

Wong, L. B., & Yeates, D. B. (1992). Luminal purinergic regulatorymechanisms of tracheal ciliary beat frequency. Am J Respir Cell Mol Biol7(4), 447−454.

Yerxa, B. R., Sabater, J. R., Davis, C. W., Stutts, M. J., Lang-Furr, M., Picher, M.,et al. (2002). Pharmacology of INS37217 [P(1)-(uridine 5′)-P(4)-(2′-deoxycytidine 5′)tetraphosphate, tetrasodium salt], a next-generation P2Y(2) receptor agonist for the treatment of cystic fibrosis. J Pharmacol Exp Ther302, 871−880.

Page 14: Relationships between cystic fibrosis transmembrane conductance regulator, extracellular nucleotides and cystic fibrosis

732 B. Marcet, J.-M. Boeynaems / Pharmacology & Therapeutics 112 (2006) 719–732

Zabner, J., Smith, J. J., Karp, P. H., Widdicombe, J. H., & Welsh, M. J. (1998).Loss of CFTR chloride channels alters salt absorption by cystic fibrosisairway epithelia in vitro. Mol Cell 2(3), 397−403.

Zimmermann, H. (1996). Extracellular purine metabolism. Drug Dev Res 39,337−352.

Zsembery, A., Boyce, A. T., Liang, L., Peti-Peterdi, J., Bell, P. D., & Schwiebert,E. M. (2003). Sustained calcium entry through P2X nucleotide receptorchannels in human airway epithelial cells. J Biol Chem 278(15),13398−13408.